1
|
Tetik-Rama S, Yilmaz-Oral D, Turkcan D, Oztekin CV, Kirlangic OF, Kirlangic FZ, Gur S. Sulfur dioxide (SO 2) donors, a new gasotransmitter, improve erectile dysfunction after castration in a rat model. Andrology 2025. [PMID: 39780452 DOI: 10.1111/andr.13839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Androgen deprivation is associated with erectile dysfunction (ED). In different animal models, sulfur dioxide (SO2) donors Na2SO3 and NaHSO3 reduced oxidative stress, fibrosis, and inflammation which contribute to the pathogenesis of androgen deprivation-induced ED, however the effect of SO2 donors on ED in castrated rats were not known. OBJECTIVE To investigate the therapeutic effect of SO2 donors, Na2SO3/NaHSO3, on ED in castrated rat model. MATERIALS AND METHODS Sprague-Dawley male rats (n = 30) were divided into four groups; control, control-treated with Na2SO3/NaHSO3, castrated, and castrated-treated with Na2SO3/NaHSO3. Castration was induced by bilateral scrotal incisions. Four weeks after castration, rats were treated with Na2SO3/NaHSO3 (0.54/0.18 mmol/kg) intraperitoneally (i.p.) for 4 weeks. Intracavernosal pressure/mean arterial pressure ratio (ICP/MAP) and total ICP were measured to evaluate in vivo erectile responses in cavernosal tissue. In vitro relaxant and contractile responses were measured in all groups. Endothelial nitric oxide synthase (eNOS), neuronal NOS (nNOS), PI3 kinase p85 alpha + gamma (PI3K), protein kinase B (AKT 1/2/3), cysteine dioxygenase-1 (CDO), and aspartate aminotransferase (AAT) expressions and localizations were evaluated by Western blotting and immunohistochemical staining. The smooth muscle/collagen ratio was evaluated by Masson's trichrome staining. RESULTS Prostate (p < 0.001) and penis weight (p < 0.001), total serum testosterone (T) level (p < 0.001), and in vivo erectile responses (p < 0.001 at 7.5 and 5 V, p < 0.05 at 2.5 V for ICP/MAP and total ICP) of castrated rats were decreased compared with control. SO2 donors improved reduced ICP/MAP ratio and total ICP (p < 0.01 at 7.5, 5, and 2.5 V for ICP/MAP and total ICP) nitrergic (p < 0.05 at 20 Hz), and endothelium-independent relaxation (p < 0.05 at 1 nM, p < 0.01 at 10 µM and 100 µM) in the castrated group. Decreased eNOS (p < 0.01) and AKT (p < 0.001) protein expressions in the castrated group were normalized by SO2. SO2 donors partially restored the reduced smooth muscle/collagen ratio in the castrated group (p < 0.001). The expressions and locations of nNOS, PI3K, CDO, and AAT proteins in penile tissue did not alter among all groups (p > 0.05). DISCUSSION AND CONCLUSION SO2 donors significantly improve erectile functions and relaxation responses in a castrated rats via ameliorating endothelial damage and fibrosis. Androgen deprivation inhibits the AKT/eNOS signaling while SO2 activates this pathway. SO2 donors may be promising for the treatment of ED in hypoandrogenic men.
Collapse
Affiliation(s)
- Seyma Tetik-Rama
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
- Department of Pharmacology, Faculty of Pharmacy, Selcuk University, Konya, Turkey
| | - Didem Yilmaz-Oral
- Department of Pharmacology, Faculty of Pharmacy, Cukurova University, Adana, Turkey
| | - Damla Turkcan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | | | - Omer Faruk Kirlangic
- Vocational School of Health Sciences, Health Service, Ankara University, Ankara, Turkey
- Department of Medical Biochemistry, Faculty of Medicine, Gazi University, Ankara, Turkey
| | | | - Serap Gur
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| |
Collapse
|
2
|
Chalise U, Hale TM. Fibroblasts under pressure: cardiac fibroblast responses to hypertension and antihypertensive therapies. Am J Physiol Heart Circ Physiol 2024; 326:H223-H237. [PMID: 37999643 PMCID: PMC11219059 DOI: 10.1152/ajpheart.00401.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 11/25/2023]
Abstract
Approximately 50% of Americans have hypertension, which significantly increases the risk of heart failure. In response to increased peripheral resistance in hypertension, intensified mechanical stretch in the myocardium induces cardiomyocyte hypertrophy and fibroblast activation to withstand increased pressure overload. This changes the structure and function of the heart, leading to pathological cardiac remodeling and eventual progression to heart failure. In the presence of hypertensive stimuli, cardiac fibroblasts activate and differentiate to myofibroblast phenotype capable of enhanced extracellular matrix secretion in coordination with other cell types, mainly cardiomyocytes. Both systemic and local renin-angiotensin-aldosterone system activation lead to increased angiotensin II stimulation of fibroblasts. Angiotensin II directly activates fibrotic signaling such as transforming growth factor β/SMAD and mitogen-activated protein kinase (MAPK) signaling to produce extracellular matrix comprised of collagens and matricellular proteins. With the advent of single-cell RNA sequencing techniques, heterogeneity in fibroblast populations has been identified in the left ventricle in models of hypertension and pressure overload. The various clusters of fibroblasts reveal a range of phenotypes and activation states. Select antihypertensive therapies have been shown to be effective in limiting fibrosis, with some having direct actions on cardiac fibroblasts. The present review focuses on the fibroblast-specific changes that occur in response to hypertension and pressure overload, the knowledge gained from single-cell analyses, and the effect of antihypertensive therapies. Understanding the dynamics of hypertensive fibroblast populations and their similarities and differences by sex is crucial for the advent of new targets and personalized medicine.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota, United States
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona, College of Medicine-Phoenix, Phoenix, Arizona, United States
| |
Collapse
|
3
|
Liu X, Zhou H, Zhang H, Jin H, He Y. Advances in the research of sulfur dioxide and pulmonary hypertension. Front Pharmacol 2023; 14:1282403. [PMID: 37900169 PMCID: PMC10602757 DOI: 10.3389/fphar.2023.1282403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 10/02/2023] [Indexed: 10/31/2023] Open
Abstract
Pulmonary hypertension (PH) is a fatal disease caused by progressive pulmonary vascular remodeling (PVR). Currently, the mechanisms underlying the occurrence and progression of PVR remain unclear, and effective therapeutic approaches to reverse PVR and PH are lacking. Since the beginning of the 21st century, the endogenous sulfur dioxide (SO2)/aspartate transaminase system has emerged as a novel research focus in the fields of PH and PVR. As a gaseous signaling molecule, SO2 metabolism is tightly regulated in the pulmonary vasculature and is associated with the development of PH as it is involved in the regulation of pathological and physiological activities, such as pulmonary vascular cellular inflammation, proliferation and collagen metabolism, to exert a protective effect against PH. In this review, we present an overview of the studies conducted to date that have provided a theoretical basis for the development of SO2-related drug to inhibit or reverse PVR and effectively treat PH-related diseases.
Collapse
Affiliation(s)
- Xin Liu
- Department of Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - He Zhou
- Departments of Medicine and Physiology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Hongsheng Zhang
- Department of Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yan He
- Department of Pediatric Cardiac Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Harris AF, Lacombe J, Sanchez-Ballester NM, Victor S, Curran KAJ, Nordquist AR, Thomas B, Gu J, Veuthey JL, Soulairol I, Zenhausern F. Decellularized Spinach Biomaterials Support Physiologically Relevant Mechanical Cyclic Strain and Prompt a Stretch-Induced Cellular Response. ACS APPLIED BIO MATERIALS 2022; 5:5682-5692. [PMID: 36368008 DOI: 10.1021/acsabm.2c00721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Recently, decellularized plant biomaterials have been explored for their use as tissue engineered substitutes. Herein, we expanded upon the investigation of the mechanical properties of these materials to explore their elasticity as many anatomical areas of the body require biomechanical dynamism. We first constructed a device to secure the scaffold and induce a strain within the physiological range of the normal human adult lung during breathing (12-20 movements/min; 10-20% elongation). Results showed that decellularized spinach leaves can support cyclic strain for 24 h and displayed heterogeneous local strain values (7.76-15.88%) as well as a Poisson's ratio (0.12) similar to that of mammalian lungs (10.67-19.67%; 0.01), as opposed to an incompressible homogeneous standard polymer (such as PDMS (10.85-12.71%; 0.4)). Imaging and mechanical testing showed that the vegetal scaffold exhibited strain hardening but maintained its structural architecture and water retention capacity, suggesting an unaltered porosity. Interestingly, we also showed that cells seeded on the scaffold can also sense the mechanical strain as demonstrated by a nuclear reorientation perpendicular to strain direction (63.3° compared to 41.2° for nonstretched cells), a nuclear location of YAP and increased expression of YAP target genes, a high cytoplasmic calcium level, and an elevated expression level of collagen genes (COL1A1, COL3A1, COL4A1, and COL6A) with an increased collagen secretion at the protein level. Taken together, these data demonstrated that decellularized plant leaf tissues have an inherent elastic property similar to that found in the mammalian system to which cells can sense and respond.
Collapse
Affiliation(s)
- Ashlee F Harris
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, Arizona85004, United States
| | - Jerome Lacombe
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, Arizona85004, United States.,Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 425 N 5th St., Phoenix, Arizona85004, United States
| | - Noelia M Sanchez-Ballester
- ICGM, CNRS, ENSCM, University Montpellier, 34000Montpellier, France.,Department of Pharmacy, Nîmes University Hospital, 30900Nîmes, France
| | - Shaun Victor
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, Arizona85004, United States
| | - Killian A J Curran
- School of Pharmaceutical Sciences, University of Geneva, Quai Ernest-Ansermet, 1211 Genève 4, Geneva, Switzerland
| | - Alan R Nordquist
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, Arizona85004, United States
| | - Baiju Thomas
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, Arizona85004, United States
| | - Jian Gu
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, Arizona85004, United States.,Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 425 N 5th St., Phoenix, Arizona85004, United States
| | - Jean-Luc Veuthey
- School of Pharmaceutical Sciences, University of Geneva, Quai Ernest-Ansermet, 1211 Genève 4, Geneva, Switzerland
| | - Ian Soulairol
- ICGM, CNRS, ENSCM, University Montpellier, 34000Montpellier, France.,Department of Pharmacy, Nîmes University Hospital, 30900Nîmes, France
| | - Frederic Zenhausern
- Center for Applied NanoBioscience and Medicine, College of Medicine Phoenix, University of Arizona, 475 North 5th Street, Phoenix, Arizona85004, United States.,Department of Basic Medical Sciences, College of Medicine Phoenix, University of Arizona, 425 N 5th St., Phoenix, Arizona85004, United States.,School of Pharmaceutical Sciences, University of Geneva, Quai Ernest-Ansermet, 1211 Genève 4, Geneva, Switzerland.,Department of Biomedical Engineering, College of Engineering, The University of Arizona, Tucson, Arizona85721, United States
| |
Collapse
|
5
|
Suki B, Bates JH, Bartolák-Suki E. Remodeling of the Aged and Emphysematous Lungs: Roles of Microenvironmental Cues. Compr Physiol 2022; 12:3559-3574. [PMID: 35766835 PMCID: PMC11470990 DOI: 10.1002/cphy.c210033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Aging is a slow process that affects all organs, and the lung is no exception. At the alveolar level, aging increases the airspace size with thicker and stiffer septal walls and straighter and thickened collagen and elastic fibers. This creates a microenvironment that interferes with the ability of cells in the parenchyma to maintain normal homeostasis and respond to injury. These changes also make the lung more susceptible to disease such as emphysema. Emphysema is characterized by slow but progressive remodeling of the deep alveolar regions that leads to airspace enlargement and increased but disorganized elastin and collagen deposition. This remodeling has been attributed to ongoing inflammation that involves inflammatory cells and the cytokines they produce. Cellular senescence, another consequence of aging, weakens the ability of cells to properly respond to injury, something that also occurs in emphysema. These factors conspire to make alveolar walls more prone to mechanical failure, which can set emphysema in motion by driving inflammation through immune stimulation by protein fragments. Both aging and emphysema are influenced by microenvironmental conditions such as local inflammation, chemical makeup, tissue stiffness, and mechanical stresses. Although aging and emphysema are not equivalent, they have the potential to influence each other in synergistic ways; aging sets up the conditions for emphysema to develop, while emphysema may accelerate cellular senescence and thus aging itself. This article focuses on the similarities and differences between the remodeled microenvironment of the aging and emphysematous lung, with special emphasis on the alveolar septal wall. © 2022 American Physiological Society. Compr Physiol 12:3559-3574, 2022.
Collapse
Affiliation(s)
- Béla Suki
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Jason H.T. Bates
- Depatment of Medicine, University of Vermont Larner College of Medicine, Burlington, Vermont
| | | |
Collapse
|
6
|
Kuiper SZ, Tol RRV, Lataster A, Cleutjens JP, Melenhorst J, Dijk PV, van Kuijk SM, Breukink SO. A Morphometric Analysis of Pathological Alterations in Hemorrhoidal Disease Versus Normal Controls: A Controlled Trial. JOURNAL OF COLOPROCTOLOGY 2022. [DOI: 10.1055/s-0041-1742257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Abstract
Objective Until today, the true pathophysiology of hemorrhoidal disease (HD) has not yet been unraveled. More and more evidence guides us towards the hypothesis that reduced connective tissue stability is associated with a higher incidence of hemorrhoids. The present study aimed to compare the quantity and quality of collagen, and vessel morphometrics, in patients with symptomatic HD compared with normal controls.
Methods Twenty-two samples of grade III and grade IV HD tissue from patients undergoing a hemorrhoidectomy between January 2004 and June 2015 were included in the study group. Samples of 15 individuals without symptomatic HD who donated their body to science and died a natural death served as controls. The quantity and quality of anal collagen, and anal vessel morphometrics were objectified. The quality of collagen was subdivided in young (immature) and old (mature) collagen.
Results Patients with HD had an increased percentage of total anal collagen (62.1 ± 13.8 versus 18.7 ± 14.5%; p = 0.0001), a decreased percentage of young collagen (0.00009 ± 0.00008 versus 0.0008 ± 0.0008%; p = 0.001), and a smaller surface area of the anal vessels (795.1 ± 1215.9 micrometre2 versus 1219.0 ± 1976.1; p = 0.003) compared with controls. The percentage of old collagen did not differ between the control and study groups (0.588 ± 0.286% versus 0.389 ± 0.242%; p = 0.06).
Conclusion The outcomes of the present study suggest that alterations in anal collagen composition may play a role in the formation of hemorrhoids.
Collapse
Affiliation(s)
- Sara Z. Kuiper
- Department of Surgery, School of Nutrition and Translation Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Robin R. van Tol
- Department of Surgery, Diakonessenhuis Medical Centre, Utrecht, The Netherlands
| | - Arno Lataster
- Department of Anatomy and Embryology, Maastricht University, Maastricht, The Netherlands
| | - Jack P.M. Cleutjens
- Department of Pathology, Maastricht University Medical Centre and School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Jarno Melenhorst
- Department of Surgery, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Paul van Dijk
- Department of Anatomy and Embryology, Maastricht University, Maastricht, The Netherlands
| | - Sander M.J. van Kuijk
- Department of Clinical Epidemiology and Medical Technology Assessment, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Stéphanie O. Breukink
- Department of Surgery, School of Nutrition and Translation Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Department of Surgery, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Surgery, School for Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
7
|
Huang Y, Zhang H, Lv B, Tang C, Du J, Jin H. Sulfur Dioxide: Endogenous Generation, Biological Effects, Detection, and Therapeutic Potential. Antioxid Redox Signal 2022; 36:256-274. [PMID: 34538110 DOI: 10.1089/ars.2021.0213] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Previously, sulfur dioxide (SO2) was recognized as an air pollutant. However, it is found to be endogenously produced in mammalian tissues. As a new gasotransmitter, SO2 is involved in regulating the structure and function of blood vessels, heart, lung, gastrointestinal tract, nervous system, etc.Recent Advances: Increasing evidence showed that endogenous SO2 regulates cardiovascular physiological processes, such as blood pressure control, vasodilation, maintenance of the normal vascular structure, and cardiac negative inotropy. Under pathological conditions including hypertension, atherosclerosis, vascular calcification, aging endothelial dysfunction, myocardial injury, myocardial hypertrophy, diabetic myocardial fibrosis, sepsis-induced cardiac dysfunction, pulmonary hypertension, acute lung injury, colitis, epilepsy-related brain injury, depression and anxiety, and addictive drug reward memory consolidation, endogenous SO2 protects against the pathological changes via different molecular mechanisms and the disturbed SO2/aspartate aminotransferase pathway is likely involved in the mechanisms for the earlier mentioned pathologic processes. Critical Issues: A comprehensive understanding of the biological effects of endogenous SO2 is extremely important for the development of novel SO2 therapy. In this review, we summarized the biological effects, mechanism of action, SO2 detection methods, and its related prodrugs. Future Directions: Further studies should be conducted to understand the effects of endogenous SO2 in various physiological and pathophysiological processes and clarify its underlying mechanisms. More efficient and accurate SO2 detection methods, as well as specific and effective SO2-releasing systems should be designed for the treatment and prevention of clinical related diseases. The translation from SO2 basic medical research to its clinical application is also worthy of further study. Antioxid. Redox Signal. 36, 256-274.
Collapse
Affiliation(s)
- Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Heng Zhang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Boyang Lv
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
8
|
Richardson WJ, Rogers JD, Spinale FG. Does the Heart Want What It Wants? A Case for Self-Adapting, Mechano-Sensitive Therapies After Infarction. Front Cardiovasc Med 2021; 8:705100. [PMID: 34568449 PMCID: PMC8460777 DOI: 10.3389/fcvm.2021.705100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
There is a critical need for interventions to control the development and remodeling of scar tissue after myocardial infarction. A significant hurdle to fibrosis-related therapy is presented by the complex spatial needs of the infarcted ventricle, namely that collagenous buildup is beneficial in the ischemic zone but detrimental in the border and remote zones. As a new, alternative approach, we present a case to develop self-adapting, mechano-sensitive drug targets in order to leverage local, microenvironmental mechanics to modulate a therapy's pharmacologic effect. Such approaches could provide self-tuning control to either promote fibrosis or reduce fibrosis only when and where it is beneficial to do so.
Collapse
Affiliation(s)
| | - Jesse D Rogers
- Department of Bioengineering, Clemson University, Clemson, SC, United States
| | - Francis G Spinale
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine and Columbia Veterans Affairs Health Care System, Columbia, SC, United States
| |
Collapse
|
9
|
Roubenne L, Marthan R, Le Grand B, Guibert C. Hydrogen Sulfide Metabolism and Pulmonary Hypertension. Cells 2021; 10:cells10061477. [PMID: 34204699 PMCID: PMC8231487 DOI: 10.3390/cells10061477] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a severe and multifactorial disease characterized by a progressive elevation of pulmonary arterial resistance and pressure due to remodeling, inflammation, oxidative stress, and vasoreactive alterations of pulmonary arteries (PAs). Currently, the etiology of these pathological features is not clearly understood and, therefore, no curative treatment is available. Since the 1990s, hydrogen sulfide (H2S) has been described as the third gasotransmitter with plethoric regulatory functions in cardiovascular tissues, especially in pulmonary circulation. Alteration in H2S biogenesis has been associated with the hallmarks of PH. H2S is also involved in pulmonary vascular cell homeostasis via the regulation of hypoxia response and mitochondrial bioenergetics, which are critical phenomena affected during the development of PH. In addition, H2S modulates ATP-sensitive K+ channel (KATP) activity, and is associated with PA relaxation. In vitro or in vivo H2S supplementation exerts antioxidative and anti-inflammatory properties, and reduces PA remodeling. Altogether, current findings suggest that H2S promotes protective effects against PH, and could be a relevant target for a new therapeutic strategy, using attractive H2S-releasing molecules. Thus, the present review discusses the involvement and dysregulation of H2S metabolism in pulmonary circulation pathophysiology.
Collapse
Affiliation(s)
- Lukas Roubenne
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut-Lévêque, F-33604 Pessac, France; (L.R.); (R.M.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ Bordeaux, U1045, 146 Rue Léo Saignat, F-33000 Bordeaux, France
- OP2 Drugs, Avenue du Haut Lévêque, F-33604 Pessac, France;
| | - Roger Marthan
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut-Lévêque, F-33604 Pessac, France; (L.R.); (R.M.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ Bordeaux, U1045, 146 Rue Léo Saignat, F-33000 Bordeaux, France
- CHU de Bordeaux, Avenue du Haut Lévêque, F-33604 Pessac, France
| | - Bruno Le Grand
- OP2 Drugs, Avenue du Haut Lévêque, F-33604 Pessac, France;
| | - Christelle Guibert
- INSERM, Centre de Recherche Cardio-Thoracique de Bordeaux, U1045, Avenue du Haut-Lévêque, F-33604 Pessac, France; (L.R.); (R.M.)
- Centre de Recherche Cardio-Thoracique de Bordeaux, Univ Bordeaux, U1045, 146 Rue Léo Saignat, F-33000 Bordeaux, France
- Correspondence:
| |
Collapse
|
10
|
Notermans T, Tanska P, Korhonen RK, Khayyeri H, Isaksson H. A numerical framework for mechano-regulated tendon healing-Simulation of early regeneration of the Achilles tendon. PLoS Comput Biol 2021; 17:e1008636. [PMID: 33556080 PMCID: PMC7901741 DOI: 10.1371/journal.pcbi.1008636] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/23/2021] [Accepted: 12/15/2020] [Indexed: 12/19/2022] Open
Abstract
Mechano-regulation during tendon healing, i.e. the relationship between mechanical stimuli and cellular response, has received more attention recently. However, the basic mechanobiological mechanisms governing tendon healing after a rupture are still not well-understood. Literature has reported spatial and temporal variations in the healing of ruptured tendon tissue. In this study, we explored a computational modeling approach to describe tendon healing. In particular, a novel 3D mechano-regulatory framework was developed to investigate spatio-temporal evolution of collagen content and orientation, and temporal evolution of tendon stiffness during early tendon healing. Based on an extensive literature search, two possible relationships were proposed to connect levels of mechanical stimuli to collagen production. Since literature remains unclear on strain-dependent collagen production at high levels of strain, the two investigated production laws explored the presence or absence of collagen production upon non-physiologically high levels of strain (>15%). Implementation in a finite element framework, pointed to large spatial variations in strain magnitudes within the callus tissue, which resulted in predictions of distinct spatial distributions of collagen over time. The simulations showed that the magnitude of strain was highest in the tendon core along the central axis, and decreased towards the outer periphery. Consequently, decreased levels of collagen production for high levels of tensile strain were shown to accurately predict the experimentally observed delayed collagen production in the tendon core. In addition, our healing framework predicted evolution of collagen orientation towards alignment with the tendon axis and the overall predicted tendon stiffness agreed well with experimental data. In this study, we explored the capability of a numerical model to describe spatial and temporal variations in tendon healing and we identified that understanding mechano-regulated collagen production can play a key role in explaining heterogeneities observed during tendon healing.
Collapse
Affiliation(s)
- Thomas Notermans
- Department of Biomedical Engineering, Lund University, Lund, Sweden
- * E-mail:
| | - Petri Tanska
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Rami K. Korhonen
- Department of Applied Physics, University of Eastern Finland, Kuopio, Finland
| | - Hanifeh Khayyeri
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Hanna Isaksson
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| |
Collapse
|
11
|
Cai H, Wang X. Effect of sulfur dioxide on vascular biology. Histol Histopathol 2020; 36:505-514. [PMID: 33319344 DOI: 10.14670/hh-18-290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Gasotransmitters, such as nitric oxide, carbon monoxide and hydrogen sulfide, can be generated endogenously. These gasotransmitters play important roles in vascular biology, including vasorelaxation and inhibition of vascular smooth muscle cell (VSMC) proliferation. In recent years, sulfur dioxide (SO₂) has been considered as a fourth gasotransmitter. SO₂ is present in air pollution. Moreover, SO₂ toxicity, including oxidative stress and DNA damage, has been extensively reported in previous studies. Recent studies have shown that SO₂ can be endogenously generated in various organs and vascular tissues, where it regulates vascular tone, vascular smooth cell proliferation and collagen synthesis. SO₂ can decrease blood pressure in rats, inhibit smooth muscle cell proliferation and collagen accumulation and promote collagen degradation, and improve vascular remodelling. SO₂ can decrease cardiovascular atherosclerotic plaques by enhancing the antioxidant effect and upregulating nitric oxide/nitric oxide synthase and hydrogen sulfide/cystathionine-γ-lyase pathways. SO₂ can also ameliorate vascular calcification via the transforming growth factor - β1/Smad pathway. The effect of SO₂ on vascular regulation has attracted great interest. SO₂ may be a novel mediator in vascular biology.
Collapse
Affiliation(s)
- Huijun Cai
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Xinbao Wang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
12
|
Abstract
Mechanotransduction, a conversion of mechanical forces into biochemical signals, is essential for human development and physiology. It is observable at all levels ranging from the whole body, organs, tissues, organelles down to molecules. Dysregulation results in various diseases such as muscular dystrophies, hypertension-induced vascular and cardiac hypertrophy, altered bone repair and cell deaths. Since mechanotransduction occurs at nanoscale, nanosciences and applied nanotechnology are powerful for studying molecular mechanisms and pathways of mechanotransduction. Atomic force microscopy, magnetic and optical tweezers are commonly used for force measurement and manipulation at the single molecular level. Force is also used to control cells, topographically and mechanically by specific types of nano materials for tissue engineering. Mechanotransduction research will become increasingly important as a sub-discipline under nanomedicine. Here we review nanotechnology approaches using force measurements and manipulations at the molecular and cellular levels during mechanotransduction, which has been increasingly play important role in the advancement of nanomedicine.
Collapse
Affiliation(s)
- Xiaowei Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Fumihiko Nakamura
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
13
|
Song Y, Peng H, Bu D, Ding X, Yang F, Zhu Z, Tian X, Zhang L, Wang X, Tang C, Huang Y, Du J, Jin H. Negative auto-regulation of sulfur dioxide generation in vascular endothelial cells: AAT1 S-sulfenylation. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30306-5. [PMID: 32087961 DOI: 10.1016/j.bbrc.2020.02.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 02/06/2020] [Indexed: 01/10/2023]
Abstract
Recently, endogenous sulfur dioxide (SO2) has been found to exert an important function in the cardiovascular system. However, the regulatory mechanism for SO2 generation has not been entirely clarified. Hence, we aimed to explore the possible auto-regulation of endogenous SO2 generation and its mechanisms in vascular endothelial cells. We showed that SO2 did not affect the protein expression of aspartate aminotransferase 1 (AAT1), a major SO2 synthesis enzyme, but significantly inhibited AAT activity in primary human umbilical vein endothelial cells (HUVECs) and porcine purified AAT1 protein. An AAT1 enzymatic kinetic study showed that SO2 reduced the Vmax (1.89 ± 0.10 vs 2.55 ± 0.12, μmol/mg/min, P < 0.05) and increased the Km (35.97 ± 9.54 vs 19.33 ± 1.76 μmol/L, P < 0.05) values. Furthermore, SO2 induced S-sulfenylation of AAT1 in primary HUVECs and purified AAT1 protein. LC-MS/MS analysis indicated that SO2 sulfenylated AAT1 at Cys192. Mechanistically, thiol reductant DTT treatment or C192S mutation prevented SO2-induced AAT1 sulfenylation and the subsequent inhibition of AAT activity in purified AAT1 protein and primary HUVECs. Our findings reveal, for the first time, a mechanism of auto-regulation of SO2 generation through sulfenylation of AAT1 at Cys192 to suppress AAT activity in vascular endothelial cells. These findings will greatly deepen the understanding of regulatory mechanisms in the cardiovascular homeostasis.
Collapse
Affiliation(s)
- Yunjia Song
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China; Research Unit of Clinical Diagnosis and Treatment of Pediatric Syncope and Cardiovascular Diseases, Chinese Academy of Medical Sciences, China
| | - Hanlin Peng
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Dingfang Bu
- Research Center, Peking University First Hospital, Beijing, 100034, China
| | - Xiang Ding
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fuquan Yang
- Key Laboratory of Protein and Peptide Pharmaceuticals & Laboratory of Proteomics, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhigang Zhu
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Xiaoyu Tian
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Lulu Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Xiuli Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Centre, Beijing, 100191, China; Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, 100191, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China; Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, 100191, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
14
|
Lopez AL, Larina IV. Second harmonic generation microscopy of early embryonic mouse hearts. BIOMEDICAL OPTICS EXPRESS 2019; 10:2898-2908. [PMID: 31259060 PMCID: PMC6583332 DOI: 10.1364/boe.10.002898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 05/15/2023]
Abstract
The understanding of biomechanical regulation of early heart development in genetic mouse models can contribute to improved management of congenital cardiovascular defects and embryonic cardiac failures in humans. The extracellular matrix (ECM), and particularly fibrillar collagen, are central to heart biomechanics, regulating tissue strength, elasticity and contractility. Here, we explore second harmonic generation (SHG) microscopy for visualization of establishing cardiac fibers such as collagen in mouse embryos through the earliest stages of development. We detected significant increase in SHG positive fibrillar content and organization over the first 24 hours after initiation of contractions. SHG microscopy revealed regions of higher fibrillar organization in regions of higher contractility and reduced fibrillar content and organization in mouse Mlc2a model with cardiac contractility defect, suggesting regulatory role of mechanical load in production and organization of structural fibers from the earliest stages. Simultaneous volumetric SHG and two-photon excitation microscopy of vital fluorescent reporter EGFP in the heart was demonstrated. In summary, these data set SHG microscopy as a valuable non-bias imaging tool to investigate mouse embryonic cardiogenesis and biomechanics.
Collapse
Affiliation(s)
- Andrew L. Lopez
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Irina V. Larina
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
15
|
Qiu C, Chen M, Yao J, Sun X, Xu J, Zhang R, Wang X, Li G, Qian S. Mechanical Strain Induces Distinct Human Scleral Fibroblast Lineages: Differential Roles in Cell Proliferation, Apoptosis, Migration, and Differentiation. Invest Ophthalmol Vis Sci 2019; 59:2401-2410. [PMID: 29847646 DOI: 10.1167/iovs.18-23855] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to explore the effect of mechanical strain on human scleral fibroblasts (HSFs) and compare cell behaviors of HSFs from distinct regions. Methods Primary HSFs were cultivated using a digestive protocol. Cells were seeded on collagen I-coated Bioflex plates, and a FX-5000 tension system was used to perform biaxial mechanical strain in vitro. We applied 10%, 0.5-Hz mechanical strain. Cell behaviors of peripapillary and periphery HSFs were compared after the strain. Edu imaging, Cell Counting Kit-8 assay, and cell cycle flow cytometry were conducted to analyze cell proliferation ability. For cell apoptosis, flow cytometry of Annexin V/propidium iodide, caspase 3 activity, and Western blot were performed. Immunofluorescence, real-time PCR, and Western blot were used to investigate cell differentiation. A migration assay was also performed. Results Under the mechanical strain of 10%, 0.5 Hz for 24 hours, the proliferation ability and cell apoptosis of peripapillary HSFs did not have a significant change. The expression of alpha-smooth muscle actin (α-SMA) slightly decreased. However, increased cell proliferation, attenuated cell apoptosis and more expression of α-SMA were shown in the periphery HSFs under the same condition. The migration rate was also increased for periphery HSFs, whereas it kept almost the same for peripapillary HSFs under 10%, 0.5-Hz strain for 8 hours. Conclusions Mechanical strain affected the cell behaviors of HSFs. The different performance of cells from distinct regions may suggest familial linages of HSFs, probably induced by mechanical strain.
Collapse
Affiliation(s)
- Chen Qiu
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China
| | - Minjie Chen
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Yao
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xinghuai Sun
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China.,State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Jianjiang Xu
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong Zhang
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Myopia, Ministry of Health, Fudan University, Shanghai, China
| | - Xin Wang
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Gang Li
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shaohong Qian
- Department of Ophthalmology, Eye and Ear, Nose, Throat Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Zhang LL, Du JB, Tang CS, Jin HF, Huang YQ. Inhibitory Effects of Sulfur Dioxide on Rat Myocardial Fibroblast Proliferation and Migration. Chin Med J (Engl) 2018; 131:1715-1723. [PMID: 29998892 PMCID: PMC6048932 DOI: 10.4103/0366-6999.235875] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Myocardial fibrosis is an important pathological change in many heart diseases, but its pathogenesis is very complex and has not yet been fully elucidated. The study was designed to examine whether endogenous sulfur dioxide (SO2) is a novel myocardial fibroblast proliferation and migration inhibitor. Methods: Primary rat myocardial fibroblasts were isolated and transfected with aspartate aminotransferase (AAT1 and AAT2) knockdown lentivirus or empty lentivirus. SO2 content in the supernatant was determined with high-performance liquid chromatography, and the expressions of AAT1, AAT2, proliferating cell nuclear antigen (PCNA), phosphorylated extracellular signal-regulated protein kinase (p-ERK), and total ERK (T-ERK) in the cells were detected. Cell migration was detected by wound healing test. Independent sample t-test (for two groups) and one-way analysis of variance (three or more groups) were used to analyze the results. Results: Both AAT1 and AAT2 knockdown significantly reduced SO2 levels (F = 31.46, P < 0.01) and AAT1/2 protein expression (AAT1, t = 12.67, P < 0.01; AAT2, t = 9.61, P < 0.01), but increased PCNA expression and Cell Counting Kit-8 (CCK-8) activity as well as the migration in rat primary myocardial fibroblasts (P < 0.01). Supplementation of SO2 rather than pyruvate significantly inhibited the increase in proliferation and migration caused by AAT knockdown (P < 0.01). Mechanistically, the ratio of p-ERK to T-ERK was significantly increased in the AAT1/2 knockdown groups compared with that in the empty lentivirus group (AAT1, t = −7.36, P < 0.01; AAT2, t = −10.97, P < 0.01). Whereas PD98059, an inhibitor of ERK activation, successfully blocked AAT knockdown-induced PCNA upregulation (F = 74.01, P > 0.05), CCK-8 activation (F = 50.14, P > 0.05), and migration augmentation in myocardial fibroblasts (24 h, F = 37.08, P > 0.05; 48 h, F = 58.60, P > 0.05). Conclusion: Endogenous SO2 might be a novel myocardial fibroblast proliferation and migration inhibitor via inhibiting the ERK signaling pathway.
Collapse
Affiliation(s)
- Lu-Lu Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Jun-Bao Du
- Department of Pediatrics, Peking University First Hospital; Division of Small Molecules and Cardiovascular Disease, Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing 100083, China
| | - Chao-Shu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Centre, Beijing 100091, China
| | - Hong-Fang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| | - Ya-Qian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
17
|
Dai J, Liu R, Zhao J, Zhang A. Sulfur dioxide improves endothelial dysfunction by downregulating the angiotensin II/AT 1R pathway in D-galactose-induced aging rats. J Renin Angiotensin Aldosterone Syst 2018; 19:1470320318778898. [PMID: 29848151 PMCID: PMC5985551 DOI: 10.1177/1470320318778898] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The aim of this study was to investigate the protective effects of sulfur dioxide (SO2) on the endothelial function of the aorta in D-galactose (D-gal)-induced aging rats. Sprague Dawley rats were randomized into a D-gal group, a D-gal + SO2 group and a control group, then injected with D-gal, D-gal + SO2 donor or equivalent volumes of saline, respectively, for 8 consecutive weeks. After 8 weeks, the mean arterial pressure was significantly increased in the D-gal group, but was lowered by SO2. SO2 significantly ameliorated the endothelial dysfunction induced by D-gal treatment. The vasorelaxant effect of SO2 was associated with the elevated nitric oxide levels and upregulated phosphorylation of endothelial nitric oxide synthase. In the D-gal group, the concentration of angiotensin II in the plasma was significantly increased, but was decreased by SO2. Moreover, levels of vascular tissue hydrogen peroxide (H2O2) and malondialdehyde were significantly lower in SO2-treated groups than those in the D-gal group. Western blot analysis showed that the expressions of oxidative stress-related proteins (the angiotensin II type 1 receptor (AT1R), and nicotinamide adenine dinucleotide phosphate oxidase subunits) were increased in the D-gal group, while they were decreased after treatment with SO2. In conclusion, SO2 attenuated endothelial dysfunction in association with the inhibition of oxidative stress injury and the downregulation of the angiotensin II/AT1R pathway in D-gal-induced aging rats.
Collapse
Affiliation(s)
- Jing Dai
- 1 Department of Clinical Diagnostics, Hebei Medical University, China
| | - Rui Liu
- 2 Department of Thoracic Surgery, Suining Central Hospital, China
| | - Jinjie Zhao
- 3 Department of Cardiovascular Surgery, Suining Central Hospital, China
| | - Aijie Zhang
- 4 Basic Laboratory, Suining Central Hospital, China
| |
Collapse
|
18
|
Yang L, Zhang H, Chen P. Sulfur dioxide attenuates sepsis-induced cardiac dysfunction via inhibition of NLRP3 inflammasome activation in rats. Nitric Oxide 2018; 81:11-20. [PMID: 30273666 DOI: 10.1016/j.niox.2018.09.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 08/16/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Sulfur dioxide (SO2) plays an important role in maintaining homeostasis of cardiovascular system. This study was aimed to investigate cardioprotective effects of SO2 on in the rat and the underlying mechanism. METHODS AND RESULTS Sepsis model induced by cecal ligation and puncture (CLP) in rats were used. SO2 donor (NaHSO3/Na2SO3, 1:3 M/M) was administered intraperitoneally at a dose of 85 mg/kg. Primary neonatal rat cardiac ventricular myocytes (NRCMs) were stimulated with LPS (1 mg/mL) in presence or absence of different concentrations of SO2 (10, 50 and 100 μmol/L). SO2 donor could restore the decreased levels of SO2 in plasma and heart of septic rats. SO2 exhibited dramatic improvement in cardiac functions. At 24 h after CLP, SO2 treatments decreased the number of TUNEL-positive cells, Bax/Bcl-2 ratio and activity of caspase-3. Moreover CLP-induced inflammatory response was also relieved by SO2. In NRCMs, SO2 could suppress the LPS-induced myocardial injury, leading to an increase in cell viability, a decrease in LDH and apoptotic rate. Western blot showed that the expression of TLR4, NLRP3, and Caspase-1 were obviously increased in myocardial tissue of CLP group or in NRCMs of LPS group, while SO2 significantly inhibited the CLP-induced or LPS-induced TLR4, NLRP3, and Caspase-1 expression. CONCLUSION SO2 attenuated sepsis-induced cardiac dysfunction likely in association with the inhibiting inflammation via TLR4/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Lin Yang
- Department of Critical Care Medicine, The First People's Hospital of Shangqiu, Shangqiu, 476100, China.
| | - Hui Zhang
- Department of Critical Care Medicine, The First People's Hospital of Shangqiu, Shangqiu, 476100, China
| | - Peili Chen
- Department of Critical Care Medicine, The First People's Hospital of Shangqiu, Shangqiu, 476100, China.
| |
Collapse
|
19
|
Richardson WJ, Kegerreis B, Thomopoulos S, Holmes JW. Potential strain-dependent mechanisms defining matrix alignment in healing tendons. Biomech Model Mechanobiol 2018; 17:1569-1580. [PMID: 30003433 DOI: 10.1007/s10237-018-1044-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/18/2018] [Indexed: 12/13/2022]
Abstract
Tendon mechanical function after injury and healing is largely determined by its underlying collagen structure, which in turn is dependent on the degree of mechanical loading experienced during healing. Experimental studies have shown seemingly conflicting outcomes: although collagen content steadily increases with increasing loads, collagen alignment peaks at an intermediate load. Herein, we explored potential collagen remodeling mechanisms that could give rise to this structural divergence in response to strain. We adapted an established agent-based model of collagen remodeling in order to simulate various strain-dependent cell and collagen interactions that govern long-term collagen content and fiber alignment. Our simulation results show two collagen remodeling mechanisms that give rise to divergent collagen content and alignment in healing tendons: (1) strain-induced collagen fiber damage in concert with increased rates of deposition at higher strains, or (2) strain-dependent rates of enzymatic degradation. These model predictions identify critical future experiments needed to isolate each mechanism's specific contribution to the structure of healing tendons.
Collapse
Affiliation(s)
- William J Richardson
- Department of Bioengineering, Clemson University, Clemson, SC, USA
- Institute for Biological Interfaces of Engineering, Clemson University, Clemson, SC, USA
| | - Brian Kegerreis
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY, USA
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville, VA, 22908, USA.
- Department of Medicine, University of Virginia, Charlottesville, VA, USA.
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
20
|
Mechanism of Mechanical Trauma-Induced Extracellular Matrix Remodeling of Fibroblasts in Association with Nrf2/ARE Signaling Suppression Mediating TGF- β1/Smad3 Signaling Inhibition. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8524353. [PMID: 29109834 PMCID: PMC5646330 DOI: 10.1155/2017/8524353] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/29/2017] [Accepted: 08/09/2017] [Indexed: 11/30/2022]
Abstract
Stress urinary incontinence (SUI) is a common hygienic problem affecting the quality of women's life worldwide. In this research, we revealed the involvement and regulation of extracellular matrix (ECM) remodeling, oxidative damage, and TGF-β1 signaling in the pathological mechanisms of mechanical trauma-induced SUI. We found that excessive mechanical strain significantly increased apoptosis rate, decreased cell viability and ECM production, and broke the balance of MMPs/TIMPs compared with the nonstrain control (NC) group. The expression levels of TGFβ1, p-Smad3, Nrf2, GPx1, and CAT were downregulated, the production of ROS, 8-OHdG, 4-HNE, and MDA was increased, and the nuclear translocation of Smad2/3 was suppressed after 5333 μstrain's treatment. Both mTGF-β1 pretreatment and Nrf2 overexpression could reverse mechanical injury-induced TGFβ1/Smad3 signaling inhibition and ECM remodeling, whereas mTGF-β1 had no effect on Nrf2 expression. Nrf2 overexpression significantly alleviated mechanical injury-induced ROS accumulation and oxidative damage; in contrast, Nrf2 silencing exhibited opposite effects. Besides, vaginal distention- (VD-) induced in vivo SUI model was used to confirm the in vitro results; Nrf2 knockout aggravates mechanical trauma-induced LPP reduction, ECM remodeling, oxidative damage, and TGF-β1/Smad3 suppression in mice. Therefore, we deduce that mechanical injury-induced ECM remodeling might be associated with Nrf2/ARE signaling suppression mediating TGF-β1/Smad3 signaling inhibition. This might reflect a new molecular target for SUI researches.
Collapse
|
21
|
Yu W, Liu D, Liang C, Ochs T, Chen S, Chen S, Du S, Tang C, Huang Y, Du J, Jin H. Sulfur Dioxide Protects Against Collagen Accumulation in Pulmonary Artery in Association With Downregulation of the Transforming Growth Factor β1/Smad Pathway in Pulmonary Hypertensive Rats. J Am Heart Assoc 2016; 5:e003910. [PMID: 27792648 PMCID: PMC5121494 DOI: 10.1161/jaha.116.003910] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 09/13/2016] [Indexed: 01/09/2023]
Abstract
BACKGROUND We aimed to explore the role of endogenous sulfur dioxide (SO2) in pulmonary vascular collagen remodeling induced by monocrotaline and its mechanisms. METHODS AND RESULTS A rat model of monocrotaline-induced pulmonary vascular collagen remodeling was developed and administered with l-aspartate-β-hydroxamate or SO2 donor. The morphology of small pulmonary arteries and collagen metabolism were examined. Cultured pulmonary arterial fibroblasts stimulated by transforming growth factor β1 (TGF-β1) were used to explore the mechanism. The results showed that in monocrotaline-treated rats, mean pulmonary artery pressure increased markedly, small pulmonary arterial remodeling developed, and collagen deposition in lung tissue and pulmonary arteries increased significantly in association with elevated SO2 content, aspartate aminotransferase (AAT) activity, and expression of AAT1 compared with control rats. Interestingly, l-aspartate-β-hydroxamate, an inhibitor of SO2 generation, further aggravated pulmonary vascular collagen remodeling in monocrotaline-treated rats, and inhibition of SO2 in pulmonary artery smooth muscle cells activated collagen accumulation in pulmonary arterial fibroblasts. SO2 donor, however, alleviated pulmonary vascular collagen remodeling with inhibited collagen synthesis, augmented collagen degradation, and decreased TGF-β1 expression of pulmonary arteries. Mechanistically, overexpression of AAT1, a key enzyme of SO2 production, prevented the activation of the TGF-β/type I TGF-β receptor/Smad2/3 signaling pathway and abnormal collagen synthesis in pulmonary arterial fibroblasts. In contrast, knockdown of AAT1 exacerbated Smad2/3 phosphorylation and deposition of collagen types I and III in TGF-β1-treated pulmonary arterial fibroblasts. CONCLUSIONS Endogenous SO2 plays a protective role in pulmonary artery collagen accumulation induced by monocrotaline via inhibition of the TGF-β/type I TGF-β receptor/Smad2/3 pathway.
Collapse
Affiliation(s)
- Wen Yu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Die Liu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Chen Liang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Todd Ochs
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Stella Chen
- Department of Biochemistry and Cellular Biology, University of California, San Diego, La Jolla, CA
| | - Selena Chen
- Department of Biochemistry and Cellular Biology, University of California, San Diego, La Jolla, CA
| | - Shuxu Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing, China Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
22
|
Role of Endogenous Sulfur Dioxide in Regulating Vascular Structural Remodeling in Hypertension. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:4529060. [PMID: 27721913 PMCID: PMC5046050 DOI: 10.1155/2016/4529060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/21/2016] [Indexed: 11/17/2022]
Abstract
Sulfur dioxide (SO2), an emerging gasotransmitter, was discovered to be endogenously generated in the cardiovascular system. Recently, the physiological effects of endogenous SO2 were confirmed. Vascular structural remodeling (VSR), an important pathological change in many cardiovascular diseases, plays a crucial role in the pathogenesis of the diseases. Here, the authors reviewed the research progress of endogenous SO2 in regulating VSR by searching the relevant data from PubMed and Medline. In spontaneously hypertensive rats (SHRs) and pulmonary hypertensive rats, SO2/aspartate aminotransferase (AAT) pathway was significantly altered. SO2 inhibited vascular smooth muscle cell (VSMC) proliferation, promoted apoptosis, inhibited the synthesis of extracellular collagen but promoted its degradation, and enhanced antioxidative capacity, thereby playing a significant role in attenuating VSR. However, the detailed mechanisms needed to be further explored. Further studies in this field would be important for the better understanding of the pathogenesis of systemic hypertension and pulmonary hypertension. Also, clinical trials are needed to demonstrate if SO2 would be a potential therapeutic target in cardiovascular diseases.
Collapse
|